The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human...

16
Mini-Review TheScientificWorldJOURNAL (2011) 11, 11531167 ISSN 1537-744X; DOI 10.1100/tsw.2011.107 *Corresponding author. ©2011 with author. Published by TheScientificWorld; www.thescientificworld.com 1153 The Origins, Specificity, and Potential Biological Relevance of Human Anti-IgG Hinge Autoantibodies Randall J. Brezski*, David M. Knight, and Robert E. Jordan Biologics Research, Centocor R&D Inc., Radnor, PA E-mail: [email protected] Received March 16, 2011; Revised May 11, 2011; Accepted May 12, 2011; Published May 26, 2011 Human anti-IgG hinge (HAH) autoantibodies constitute a class of immunoglobulins that recognize cryptic epitopes in the hinge region of antibodies exposed after proteolytic cleavage, but do not bind to the intact IgG counterpart. Detailed molecular characterizations of HAH autoantibodies suggest that they are, in some cases, distinct from natural autoantibodies that arise independent of antigenic challenge. Multiple studies have attempted to define the specificity of HAH autoantibodies, which were originally detected as binding to fragments possessing C-terminal amino acid residues exposed in either the upper or lower hinge regions of IgGs. Numerous investigators have provided information on the isotype profiles of the HAH autoantibodies, as well as correlations among protease cleavage patterns and HAH autoantibody reactivity. Several biological functions have been attributed to HAH autoantibodies, ranging from house- cleaning functions to an immunosuppressive role to restoring function to cleaved IgGs. In this review, we discuss both the historic and current literature regarding HAH autoantibodies in terms of their origins, specificity, and proposed biological relevance. KEYWORDS: autoantibodies, antibody-dependent cellular cytotoxicity, complement, monoclonal antibodies INTRODUCTION A long-standing question in immunology is how individuals are capable of generating a very broad range of diversity against foreign antigens, while at the same time eliminating reactivity against self-antigens. Autoreactive B cells are regulated by tolerance checkpoints during B-cell development, which can occur in the bone marrow (central tolerance) or in the periphery (peripheral tolerance). B cells recognize antigen through their B-cell antigen receptors (BCR), which can subsequently be secreted as antibodies into the serum as a part of the humoral immune response. When a breakdown in either B-cell central or peripheral tolerance occurs, individuals can generate autoantibodies directed against self-antigens. In some cases, autoantibodies are thought to correlate with pathological conditions attributed to autoimmune dysfunction, such as autoimmune hemolytic anemia, myasthenia gravis, and idiopathic thrombocytopenic purpora (ITP). However, most healthy individuals have circulating B cells that recognize numerous self- antigens. Several types of autoantibodies have been characterized, including those directed against

Transcript of The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human...

Page 1: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Mini-Review TheScientificWorldJOURNAL (2011) 11, 1153–1167 ISSN 1537-744X; DOI 10.1100/tsw.2011.107

*Corresponding author. ©2011 with author. Published by TheScientificWorld; www.thescientificworld.com

1153

The Origins, Specificity, and Potential Biological Relevance of Human Anti-IgG Hinge Autoantibodies

Randall J. Brezski*, David M. Knight, and Robert E. Jordan

Biologics Research, Centocor R&D Inc., Radnor, PA

E-mail: [email protected]

Received March 16, 2011; Revised May 11, 2011; Accepted May 12, 2011; Published May 26, 2011

Human anti-IgG hinge (HAH) autoantibodies constitute a class of immunoglobulins that recognize cryptic epitopes in the hinge region of antibodies exposed after proteolytic cleavage, but do not bind to the intact IgG counterpart. Detailed molecular characterizations of HAH autoantibodies suggest that they are, in some cases, distinct from natural autoantibodies that arise independent of antigenic challenge. Multiple studies have attempted to define the specificity of HAH autoantibodies, which were originally detected as binding to fragments possessing C-terminal amino acid residues exposed in either the upper or lower hinge regions of IgGs. Numerous investigators have provided information on the isotype profiles of the HAH autoantibodies, as well as correlations among protease cleavage patterns and HAH autoantibody reactivity. Several biological functions have been attributed to HAH autoantibodies, ranging from house-cleaning functions to an immunosuppressive role to restoring function to cleaved IgGs. In this review, we discuss both the historic and current literature regarding HAH autoantibodies in terms of their origins, specificity, and proposed biological relevance.

KEYWORDS: autoantibodies, antibody-dependent cellular cytotoxicity, complement, monoclonal antibodies

INTRODUCTION

A long-standing question in immunology is how individuals are capable of generating a very broad range

of diversity against foreign antigens, while at the same time eliminating reactivity against self-antigens.

Autoreactive B cells are regulated by tolerance checkpoints during B-cell development, which can occur

in the bone marrow (central tolerance) or in the periphery (peripheral tolerance). B cells recognize antigen

through their B-cell antigen receptors (BCR), which can subsequently be secreted as antibodies into the

serum as a part of the humoral immune response. When a breakdown in either B-cell central or peripheral

tolerance occurs, individuals can generate autoantibodies directed against self-antigens. In some cases,

autoantibodies are thought to correlate with pathological conditions attributed to autoimmune

dysfunction, such as autoimmune hemolytic anemia, myasthenia gravis, and idiopathic thrombocytopenic

purpora (ITP). However, most healthy individuals have circulating B cells that recognize numerous self-

antigens. Several types of autoantibodies have been characterized, including those directed against

Page 2: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1154

immunoglobulins. Humans have five classes of immunoglobulins: IgM, IgD, IgG, IgA, and IgE. The best

characterized anti-immunoglobulin autoantibodies are those directed against the Fc region of IgG, known

as rheumatoid factor, or those directed against the variable regions, known as anti-idiotype

autoantibodies. Over the years, another class of anti-immunoglobulin autoantibody has been described:

those that bind specifically to proteolytically exposed epitopes containing C-terminal amino acid residues

in either the upper or lower hinge region. These human antihinge (HAH) autoantibodies generally are not

detected as binding to the intact IgG counterpart.

The origin of HAH autoantibodies is not entirely clear. They are sometimes referred to as natural

autoantibodies (NAbs). There are at least two broad definitions attributed to the term NAbs. One

definition indicates that they arise independent of an immune response[1] and contain variable regions

that are comprised of germline-encoded genes[2]. The other definition refers to the subset of low-affinity

(and sometimes polyreactive) autoantibodies that are present at a steady-state level in otherwise healthy

individuals[3]. Variable region cloning and analysis could provide some clues as to the origin of antihinge

autoantibodies. To our knowledge, only two antihinge autoantibodies have been cloned. The heavy-chain

variable regions of those two HAH autoantibodies had 88% homology with the nearest germline

sequence[4], suggesting that somatic hypermutation had occurred during an immune response[5]. More

recent studies have shown that HAH autoantibodies exist that react with a wide range of epitopes[6],

suggesting that the two cloned antihinge autoantibodies may not be fully representative of the entire

repertoire of HAH autoantibodies.

The biological function of autoantibodies directed against cleaved IgGs is enigmatic. The presence of

antihinge-specific autoantibodies was first detected in rabbits in the 1960s when serum factors were

described that bound to determinants exposed following the cleavage of polyclonal rabbit autoantibodies

with either papain or pepsin[7,8]. Subsequent studies in the human system not only uncovered similar

specificities, but were extended to investigations of their presence in a number of disease states, including

cold agglutination (CA)[9], human immunodeficiency virus (HIV)[10], rheumatoid arthritis[11], and

systemic lupus erythematosus (SLE)[12]. Several investigations suggested that HAH autoantibodies could

play an immunosuppressive role by binding to the hinge region of antigen-engaged B cells and inducing

apoptosis via coaggregation of the BCR with FcγRIIb (reviewed in Terness et al.[5]). Other proposed

functions include complement amplification[13] or providing a surrogate Fc domain to cleaved IgGs to

restore Fc-mediated effector functions, such as antibody-dependent cellular cytotoxicity (ADCC) and

complement-dependent cytotoxicity (CDC)[6].

In this review, we will discuss each of the above-mentioned aspects of HAH autoantibodies, spanning

the early literature to the more recent findings. Because a substantial fraction of the literature reviewed is

historic, and cellular and molecular biological techniques have advanced considerably, we will also point

out where new approaches could potentially shed light on the origins, specificity, and potential biological

functions of HAH autoantibodies.

B-CELL DEVELOPMENT AND AUTOANTIBODY GENERATION

In mice, there are at least three unique B-cell lineages: B1a, B1b, and B2[14]. B1a B cells develop from

precursors in the fetal liver and are thought to contribute to natural IgM antibodies found in the serum.

The variable regions of B1a B cells are nonmutated. B1b B cells originate from precursors in the bone

marrow. In contrast to B1a B cells, B1b B cells can express mutated IgG variable regions[15]. Both B1a

and B1b B cells reside in the pleural and peritoneal cavities, are capable of responding to T-cell–

independent antigens, and participate in the first line of defense against foreign pathogens[16]. B2 B cells

develop from precursors in the bone marrow throughout life; they are capable of responding to T-cell–

dependent antigens and are considered an essential part of the adaptive immune response. At present, B1

B cells are less well characterized in humans[1,17].

In both humans and mice, B-cell development is characterized by an ordered and sequential assembly

of the BCR, beginning with assembly of the preBCR followed by the mature BCR. The mature BCR is a

Page 3: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1155

multisubunit, transmembrane protein consisting of an immunoglobulin heavy chain (IgHC) covalently

linked to an immunoglobulin light chain (IgLC) by disulfide bonds. The IgHC/IgLC heterodimer is

noncovalently linked to the signaling subunits Igα and Igβ. Expression of the IgHC is primarily regulated

by a process known as V(D)J recombination, which also contributes to the initial generation of BCR

diversity. During this process, gene segments known as the variable (V), diversity (D), and joining (J)

regions recombine to form a series of unique, heavy-chain variable regions. Expression of the IgLC is

regulated by a similar process involving recombination of the light-chain V and J gene segments. These

processes result in the broad diversity of antigen recognition that allows B cells to cope with the myriad

of foreign invaders encountered over the lifetime of the host. One potentially problematic consequence of

IgHC and IgLC recombination is that the gene rearrangements occur indiscriminately, meaning that

antigen recognition sites can recognize both foreign antigens and self-antigens. It was estimated that early

immature B cells have approximately 75% self-reactive BCRs, and those BCRs were polyreactive against

antigens such as insulin, DNA, and lipopolysaccharides (LPS)[18]. Additionally, many of the self-

reactive BCRs recognized antigens from the nucleus[18]. Most of these self-reactive B cells were

eliminated in the bone marrow by the first tolerance checkpoint. Three main processes have been

identified that are responsible for eliminating self-reactive B cells. First, bone marrow and early-stage

peripheral autoreactive B cells that recognize self-antigen can be deleted by apoptosis, a process known as

negative selection[19,20]. Second, autoreactive B cells that receive intermediate signals can undergo

additional recombination events, known as receptor editing[21]. Third, self-reactive B cells can enter a

state of unresponsiveness, termed anergy[22]. It was estimated that the majority of self-reactive cells are

eliminated by receptor editing[23].

One way in which B cells can escape peripheral tolerance is during the natural progression of an

immune response[24]. Mature, naive B cells that initially become activated in an immune response are

thought to undergo several different fate paths[25,26,27]. A subset of antigen-challenged B cells rapidly

differentiates into short-lived immunoglobulin-secreting plasma cells (PC), which contribute to the initial

defense against foreign pathogens. Another subset of activated B cells migrates to B-cell follicles to

produce germinal centers, which aids in the development of serological memory by production of

memory B cells and long-lived PC. One process that contributes to humoral memory is class-switch

recombination, whereby the immunoglobulin constant regions switch to other immunoglobulin isotypes,

such as IgG. Another process is somatic hypermutation, which entails introduction of random gene

modifications within the IgHC and IgLC variable regions. As a result of these two processes, B cells with

new variable regions and isotypes are selected within the germinal center reaction. A recent study has

indicated that a significant increase in self-reactive IgG+ memory B cells emerge out of an immune

response, suggesting that de novo autoreactive B cells can result from the process of somatic

hypermutation[28].

ORIGINS OF HAH AUTOANTIBODIES

The mechanism that results in the production of HAH autoantibodies is not entirely clear and any

association with chronological age has not been studied to our knowledge (i.e., their presence or absence

in young children). However, the presence of HAH autoantibodies can be detected to varying degrees in

most healthy adults[6]. Therefore, it seems likely that individuals expressing HAH autoantibodies could

have populations of circulating B cells in the periphery that have HAH-reactive BCRs. In investigations

on the antigen binding domain of HAH autoantibodies, Welschof and colleagues provided data to confirm

this supposition. The authors constructed a phagemid library from the peripheral blood mononuclear cells

(PBMCs) isolated from an individual that had high anti-F(ab')2 titers and were able to clone out two anti-

F(ab')2 autoantibodies[4]. The study did not evaluate the cell surface marker phenotype of the anti-

F(ab')2–expressing B cells from which the phagemid library was generated. However, the cloned variable

regions provided clues about the origin of those two anti-F(ab')2 autoantibodies. The two IgHC variable

regions had 88% homology with the closest germline sequences. In contrast, the two IgLC variable

Page 4: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1156

regions were nearly identical to the closest germline sequences (100 and 99.3% homology). Due to the

fact that the two cloned IgHC variable regions from anti-F(ab')2–expressing B cells had 88% homology

with the closest germline regions, it seems unlikely that these clones derived from NAb-expressing B

cells. As previously noted, two hallmarks of memory B cells are class-switch recombination and somatic

hypermutation, characteristics displayed by both of the cloned anti-F(ab')2 autoantibodies. Therefore, it is

possible that these two clones were derived from memory B cells.

Perhaps additional clues into the origins of HAH B cells can be inferred from the isotype of HAH

autoantibodies. A recent study conducted in our lab indicated that HAH autoantibodies display a diverse

number of immunoglobulin isotypes[6]. We found HAH autoantibodies of the IgM, IgG1, IgG2, IgG3,

and IgA isotypes, with a particular emphasis on the IgG3 isotype. The prevalence of HAH autoantibodies

of the IgG3 isotype has been noted by other labs as well[11]. Although class-switch recombination and

somatic hypermutation have been documented to occur outside of germinal centers[29,30], it is possible

that HAH autoantibodies with isotypes other than IgM could have occurred as a result of a germinal

center reaction. It is also important to note that a significant component of HAH autoantibodies was found

to be of the IgM isotype, which could perhaps represent antihinge NAbs. Taken together, since HAH

autoantibodies have been identified that react with a wide range of IgG hinge epitopes with C-terminal

exposed amino acid residues, and contain a number of different isotypes, HAH autoantibodies could

potentially develop from several distinct B-cell lineages. Therefore, the overall repertoire of HAH

autoantibodies could include NAbs as well as independent HAH autoantibody clones that developed from

an adaptive immune response.

The advent of multiparameter flow cytometric analysis has afforded investigators the ability to not

only isolate antigen-specific B cells, but also to phenotype the lineage of those B cells by analyzing cell

surface markers. Additionally, the use of single-cell sorting technologies has proven to be a useful tool for

the cloning of antigen-specific B-cell variable regions[31]. Future studies in which the variable regions of

HAH autoantibodies can be cloned, combined with cell surface marker phenotype analyses, will lead to

further insights into the genetic diversity of this population of autoantibodies.

SPECIFICITY OF HAH AUTOANTIBODIES

Following the demonstration that rabbit IgGs could be proteolytically fragmented by papain to yield Fab

fragments[32] and by pepsin to yield F(ab')2[33], it was observed that new antigenic determinants were

exposed that were recognized by pre-existing autoantibodies in rabbit serum[7,8]. Moreover, different

populations of rabbit autoantibodies that recognized Fab and F(ab')2, but that showed no similar reactivity

to intact rabbit IgG, were found. These insightful early observations (obtained with polyclonal rabbit

antigen fragments and autoantibodies, and with the agglutination methods then employed) prompted

similar human investigations.

The utility of papain and pepsin was applied to the generation of Fab and F(ab')2 fragments from

human IgGs. Two Fab fragments are released from IgG when the heavy chains are each cleaved in the

upper hinge, an event that simultaneously generates an Fc fragment in which the two heavy-chain

components remain covalently attached through the disulfide linkages of the core hinge (e.g., with

papain). F(ab')2 fragments result from two separate peptide bond cleavages in the lower hinge, leaving the

two Fab domains linked through the core disulfides (e.g., with pepsin). As in the rabbits, human serum

autoreactivity to the IgG fragments was readily detected and designated as papain agglutinators[34,35,36]

or pepsin agglutinators[37] by some authors to denote their capacities for immunoprecipitation of the

respective fragments. This topic was actively investigated, and numerous additional groups contributed to

the characterization and specificity of human antifragment autoantibodies[35,38,39,40]. Investigations

were also performed on IgGs cleaved with different enzyme sources. Bacterial proteases (subtilisin,

bromelain), human digestive enzymes (trypsin, pancreatic elastase, and chymotrypsin, in addition to

pepsin), plant-derived proteases (ficin and papain), and the human fibrinolytic protease, plasmin, were all

found to yield IgG fragments that were recognized by apparently distinct populations of human serum

Page 5: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1157

antibodies[41,42,43]. A consensus emerged that antifragment reactivity was widespread in the human

population[44,45], although of generally low affinity[46] and with considerable heterogeneity in titer

among different individuals[46,47]. It was commonly observed that an individual’s antifragment

autoantibodies showed more reactivity to F(ab')2 than to Fab fragments[38,48].

The majority of the early investigations focused on papain- and pepsin-digested polyclonal IgGs, and

these sometimes led to conflicting interpretations regarding whether autoantibody antifragment reactivity

was directed to hinge epitopes or was directed to idiotypic or framework determinants[35,49]. Even as

methodologies transitioned to more sensitive radioimmunoassays (RIA)[45,49] and enzyme-linked

immunosorbent assay (ELISA) formats[36,47], ambiguities regarding binding specificity remained due to

the polyclonal nature of the reagents employed. Indeed, one study employing epitope-mapping

approaches concluded that certain epitopes for autoantibody recognition resided within the variable and

constant regions of λIgLCs[50]. We are not aware of other studies that point to IgLC epitopes for

autoantibody recognition. Concerns about the physiological relevance of autoantibodies directed to new

antigenic epitopes generated with papain and pepsin derived from the nature of these enzymes

themselves. Papain (vegetable derived) and pepsin (active in acidic stomach conditions) do not normally

reach human nondigestive tissues or circulation. However, the peptide bonds cleaved by papain and

pepsin might reflect parallel specificities with other, more physiologically relevant, proteases to thereby

reveal analogous epitopes for autoantibody recognition. However, early detailed correlations of

autoantibody binding and cleavage site specificity were seldom possible, due at least in part to the

polyclonal and multi-isotype nature of human IgG preparations. Nevertheless, the improved assay formats

did afford better descriptions of human antifragment autoantibodies, and prompted a number of queries

regarding associations to human immune responses and pathologies (described earlier in this review). As

also mentioned previously, some studies pointed to unexpected serological findings, including that

antihinge autoantibodies were biased toward minor isotypes, including IgG3[11].

Recent investigations of antihinge autoantibodies have been facilitated by the use of monoclonal

antibodies and peptide analogs of specific hinge sequences, as well as the deployment of physiologically

relevant proteases for generation of the IgG fragments. The earlier studies in almost all cases treated Fab

and F(ab')2 as generic entities without a recognition that subtly different fragment lengths result from

different protease cleavage sites. We and others found that a number of human and bacterial proteases

cleaved human IgG1 within the upper and lower hinge domains[51,52,53,54] and defined a number of

different cleavage points therein (Fig. 1). Almost without exception, protease cleavage of IgGs has been

assessed in solution in purified systems, a set of conditions that may not always be analogous to those in

vivo. In any case, it became evident that the different proteolytically generated Fab and F(ab')2 fragments

elicited varying degrees of autoantibody recognition, and in some cases, the reactivity was higher than

that seen with the fragments generated with papain and pepsin[6].

With many of the human physiologic proteases, IgG cleavage was observed to proceed more slowly

compared to papain and pepsin. However, the natural roles of papain (defense against fungal or microbial

attack on the papaya plant) and pepsin (rapid digestion of protein in the stomach) are likely

unrepresentative of the actions of extracellular proteases in human tissue. Moreover, the human

extracellular proteases that have been described to cleave IgGs are better recognized for their natural roles

in physiology or pathology (e.g., the metalloproteinases from immune cells and tumor cells work to break

down extracellular matrix components during migration and invasion[55,56]). Nevertheless, a number of

such proteases have now been shown to cleave IgG and, in many cases, with well-defined specificity

within the hinge. The generally slower rates of solution-phase IgG cleavage compared to papain and

pepsin may have hindered appreciation for the potential impact of physiological IgG-degrading enzymes

in pathological settings. One bacterial protease, IdeS from Streptococcus pyogenes, is rapid acting and is

remarkably specific for cleavage of the IgG lower hinge[54]. Taken together, the cumulative evidence led

us to further speculate that host immune defenses against invasive cells (tumors, bacteria) may be

compromised by local protease expression[57].

Page 6: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1158

FIGURE 1. A schematic representation of a human IgG1 antibody (left side) and the amino acid sequence of the

hinge region (right side). The numbers below the amino acid sequence indicate specific protease cleavage points

within the hinge region.

To test whether endogenous proteolysis of IgG could be correlated with autoantibody reactivity to

revealed epitopes containing C-termini, it was desirable to obtain independent assessments of binding to

different potential positions of cleavage within the hinge. Peptide analog approaches have been variously

adopted to define the fine specificity of antihinge antibodies[6,48,58,59]. In one study[48], human serum

antihinge antibodies were prepared by affinity isolation on F(ab')2 fragments generated with pepsin (the

cleavage site in IgG1 assigned between L234 and L235)[48,60,61,62]. The autoantibodies were tested for

binding to two different two-chain peptide analogs, terminating in the IgG1 lower hinge “TCPPCPAP”

and the longer “TCPPCPAPELLGG” (note that the autoantibodies were isolated on the basis of their

affinity to an IgG fragment with a C-terminal cleavage site intermediate to the termini of these two

peptides). The test peptides were dimeric (in accord with the actual hinge in intact IgG1), possessed the

core disulfide-linked hinge structure, and displayed unmodified N- and C-termini. ELISA binding of

human serum samples showed strong binding to the longer dimeric peptide and less to the shorter (and

almost no binding to the single-chain peptide). The authors concluded that the polyclonal autoantibody

preparations bound to a conformational epitope requiring the (disulfide-linked) core hinge.

Subsequent studies from the same group advanced their interpretation of the core hinge being a

component of a discontinuous, conformational epitope for antihinge autoantibody binding. As previously

noted, Welschof and colleagues cloned two HAH antibodies with affinity for human F(ab')2 fragments

(pepsin generated)[4]. The two antibodies were expressed as single-chain Fv constructs and were

extensively characterized with regard to binding to dimeric hinge peptide analogs in which each position

(except cysteines) was substituted by every other amino acid[58]. The cloned autoantibodies bound to the

wild-type hinge sequence and exhibited tolerance for many substitutions in most hinge positions, with the

exception of three proline residues that appeared essential to the binding by these scFvs. It should be

noted that the double-stranded peptides could be considered analogs of proteolytically generated Fc

fragments with a free N-terminus at T225 in the upper hinge, a departure from earlier peptide studies

incorporating a free C-terminus or the more historical approaches using Fab or F(ab')2 fragment analogs

with free C-termini. Nevertheless, the results were consistent with the core hinge, constituting a

component of the epitope for these particular autoantibodies.

Studies from our own laboratory focused on HAH autoantibodies that bound to hinge structures

conforming to epitopes expected to be present on Fab and F(ab')2 fragments. We adopted an epitope-

mapping approach in which the selectivity of autoantibody binding was presumed to reflect differences in

the carboxy terminus of the proteolytically generated fragments[6]. To address this, progressive single-

chain, 14-mer peptides with an N-terminal biotin adduct and with free C-termini at each position between

D221 and L251 were individually captured on streptavidin-coated microtiter wells in ELISA. We

Page 7: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1159

confirmed that reactive autoantibodies were often IgG3s, a finding that focused attention on this isotype

for the subsequent investigations[6]. IgG3 autoantibodies from human serum showed a discontinuous

pattern of binding reactivity for peptides with different C-termini in the IgG1 hinge[6]. First, no

autoantibody reactivity was found for peptides terminating within the core “CPPC” sequence, suggesting

that this component was either not sufficient to define an epitope or was comprised with amino acid

positions that do not normally get exposed as antigens. Moderate levels of IgG binding were directed to

peptide analogs in the upper hinge with termini at K222, T223, and H224 (analogs of sites of cleavage by

plasmin, neutrophil elastase [greatest reactivity], and papain, respectively). Contrastingly, robust

autoantibody binding to lower hinge analogs with C-terminal positions between P232 and F241 was

found. This sequence encompasses points of cleavage by several human metalloproteinases, human

cathepsin G, (porcine) pepsin, the Staphylococcus aureus GluV8 protease, and the Streptococcus

pyogenes IdeS protease[53]. Antigenic peptides with intervening termini that are not identified as sites of

protease cleavage also exist within this sequence and could represent sites of overlapping specificities to

adjacent, known protease sites or may correspond to cleavage sites by yet unidentified enzymes.

It was noted that single-chain 14-mer peptide analogs of the same hinge region, but containing each

position as a free N-terminus (and with the C-terminus blocked), yielded negligible autoantibody

binding[63]. Thus, peptides with a particular amino acid as a C-terminus were often recognized by

autoantibodies, whereas peptides originating from the opposite direction with the same amino acid as a

free N-terminus were not bound. The reason for this disparity in antigenicity at the same site, but with

opposed directionality, is unclear. Other investigators found a class of autoantibodies with specificity for

a conformational hinge epitope that incorporated the disulfide-linked core and without regard to the

orientation of the cleavage point[48]. We did not incorporate the core hinge in our epitope-mapping

strategy, but instead showed specificity for the C-terminus of the single-chain analogs of cleavage sites.

These differences may be revealing in pointing out the potential for multiple autoimmune specificities.

Proteolysis of IgGs with a number of physiological proteases yielded intermediate products not

usually observed with more rapidly acting papain and pepsin, and therefore not usually encountered in

laboratory digestion reactions. Specifically, human and bacterial extracellular proteases have been shown

to cleave IgG to F(ab')2 in two steps[52,53,64,65,66]. The first step, to a single-cleaved intermediate in the

lower hinge (without removal of the Fc region), was found to be more rapid in vitro than the second

step[66], and single-cleaved intermediates were observed to accumulate in pathological settings in

vivo[53,66]. The limited proteolysis and accumulation of intermediate digestion products in vivo may be

relevant with regard to recent observations that single-cleavage versions of cell-targeting IgGs lose

substantial, if not all, Fc-mediated effector functions related to target cell killing[66], and that the Fc-

mediated functions of such species may be restored by the binding of HAH antibody preparations to cell-

bound cleaved IgGs[6]. Single-clipped hinge domains of IgG (and perhaps especially those that remain

bound to cell surfaces) might expose physiologically relevant epitopes to allow host immune recognition

of otherwise cryptic hinge elements. It remains to be shown if physiological IgG cleavage can be

correlated with autoantibody reactivity to exposed antigens. Also remaining to be investigated are any

clinical effects or consequences to patients with autoimmune diseases following the administration of

high doses of intravenous immunoglobulin preparations containing low, but detectable (<0.1%), amounts

of these autoantibodies[6].

BIOLOGY OF HAH AUTOANTIBODIES

Autoantibodies have been studied for decades and have been linked to normal, immunological, regulatory

mechanisms, as well as to a variety of diseases, including autoimmune disorders, such as SLE, multiple

sclerosis (MS), nephritis, and others. Autoantibodies whose targets are immunoglobulins comprise one

class of autoantibodies that has been studied extensively. For convenience, these anti-immunoglobulin

autoantibodies are often classified according to the domain of the antibody molecule recognized by the

autoantibodies. Anti-immunoglobulins may target the Fc constant region (e.g., rheumatoid factors), or the

Page 8: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1160

Fab or F(ab')2 domains. Autoantibodies that recognize the Fab or F(ab')2 domains can be further divided

into anti-idiotypic antibodies that target the variable regions, and antibodies that target the constant

regions (heavy-chain CH1, hinge, IgLCκ or λ) of the Fab or F(ab')2 domains. The proteolytic

methodologies cited above were developed to cleave antibody molecules and separate the domains, thus

facilitating the characterization of anti-immunoglobulins. As detailed, HAH autoantibodies react with Fab

or F(ab')2 fragments of immunoglobulins (primarily IgGs), but not with the intact IgG molecules.

Because, as discussed above, Fab or F(ab')2 fragments contain many potential autoantibody targets other

than amino acids exposed by proteolytic cleavage, attribution of biological properties to HAH

autoantibodies must be contingent upon experimental demonstration that the hinge region is, in fact, the

autoantibody target.

STUDIES WITH ANTI-FAB/ANTI-F(AB')2 AUTOANTIBODIES

Anti-Fab and anti-F(ab')2 autoantibodies exist in most normal humans[67]. Early speculation on the

possible role of these normal autoantibodies centered around clearance mechanisms to remove

proteolyzed antibodies in a normal process of antibody catabolism[46], although such a house-cleaning

role has not been directly demonstrated. Anti-Fab or anti-F(ab')2 autoantibodies have been associated with

a variety of diseases. Persselin and Stevens[11] studied anti-Fab antibodies in the sera of rheumatoid

arthritis patients and found that the autoantibodies were generally elevated compared to normal sera and

were predominantly minor isotypes (IgG3 and IgG4). The prevalence of minor isotypes is consistent with

a number of other studies[6,10], but its biological significance remains to be delineated. Anti-F(ab')2

autoantibodies were studied in CA disease, which is characterized by the presence of autoantibodies

frequently directed against red blood cell (RBC) antigens[9]. An inverse correlation was noted between

RBC autoantibodies and anti-F(ab')2 autoantibodies; the authors argue for an important role of the anti-

F(ab’)2 autoantibodies in the regulation of RBC autoantibody production in CA patients. In another

disease associated with elevated autoantibodies, SLE, a similar inverse correlation was noted; in this case,

between anti-DNA autoantibodies and anti-F(ab')2 autoantibodies[12]. Patients with active SLE often

have low serum anti-F(ab')2 and patients with SLE in remission often have high levels of serum anti-

F(ab')2 autoantibodies. The authors suggested that serum anti-F(ab')2 may be depleted as the result of

increased anti-F(ab')2–containing immune complex deposition in renal tissues. Consistent with this idea,

enrichment of anti-F(ab')2 autoantibodies was observed in renal biopsy tissues from patients with active

SLE. Anti-Fab autoantibodies have also been studied in patients with acquired immunodeficiency

syndrome (AIDS) and aids-related complex (ARC)[68]. Patients with AIDS/ARC had significantly higher

anti-Fab autoantibodies than HIV-positive patients without AIDS/ARC or uninfected control subjects. A

highly significant inverse association was found between anti-Fab autoantibodies and CD4+ T-cell

counts. An association has also been observed between IgA anti-Fab autoantibodies and disease stage in

head-and-neck cancer patients[69]. Patients with squamous-cell carcinoma (SCCHN) and adenoid cystic

carcinoma (ACCHN) of the head and neck had higher levels of IgA anti-Fab autoantibodies than healthy

controls. In patients with SCCHN, there was a direct correlation between disease stage and IgA anti-Fab

activity. A strong association was also seen between anti-Fab autoantibodies and severe infections by

various microorganisms[67]. Anti-Fab autoantibodies were highest in patients with septicemia due to

Gram-positive cocci.

The studies noted above describe correlations between various diseases and anti-Fab or anti-F(ab’)2

autoantibodies, with little data addressing mechanisms to explain the role of autoantibodies in the

initiation or maintenance of disease. Because precise epitope-mapping information on the targets of anti-

Fab and anti-F(ab')2 autoantibodies and specific monoclonal reagents were not yet available, these early

correlative studies could not distinguish among the different categories of anti-Fab or anti-F(ab')2

autoantibodies, so that any involvement of specific HAH autoantibodies in these reported studies was

unclear.

Page 9: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1161

In addition to disease associations, anti-F(ab')2 autoantibodies have been implicated in immune

regulatory mechanisms. In a series of reports, Terness and colleagues demonstrated a potential

immunoregulatory role of human anti-F(ab')2 autoantibodies[5]. They focused on nonidiotypic

autoantibodies that could potentially react with various parts of the F(ab')2 molecule, including the IgLC

constant region, the CH1 IgHC region, and the hinge region. An Fc-dependent immunosuppressive

function of the autoantibodies was demonstrated, and it was proposed that the autoantibodies may

contribute to the termination of a successful antibody response and/or the control of autoreactive B-cell

clones. One suggested mechanism for the immunosuppressive effect involves binding of IgG anti-F(ab')2

to the immunoglobulin-containing membrane BCR via the autoantibody Fab region, followed by cross-

linking of the BCR and the inhibitory FcγRIIb receptor on the surface of B cells via the Fc region of the

anti-F(ab')2 autoantibody. Cross-linking the BCR and FcγRIIb can lead to apoptosis of the B cell,

resulting in an immunosuppressive effect (Fig. 2A). In the simplest version of this model, the anti-F(ab')2

targets on the membrane immunoglobulin molecule are constant-region determinants accessible on the

immunoglobulin surface (e.g., IgHC CH1 or IgLCλ or κ). For HAH autoantibodies (whose targets are

exposed upon proteolytic cleavage of IgG) to mediate suppression in this model, there must be a

mechanism to reveal HAH epitopes on the membrane immunoglobulin since HAH autoantibodies do not

bind to intact immunoglobulin. To accommodate the possibility of HAH involvement, the authors

postulate that upon antigen binding to the Fab region of the membrane immunoglobulin portion of the

BCR, a conformational change is transmitted to the hinge region, exposing specific HAH epitopes and

allowing HAH binding with subsequent cross-linking to FcγRIIb. Consistent with this idea, selective

inactivation of B cells has been shown when the BCR is occupied with antigen[70]. These studies showed

that anti-immunoglobulin antibodies can suppress B-cell antibody production in an Fc-dependent manner,

although it was not shown directly that HAH autoantibodies alone can mediate the suppression. A direct

role for HAH autoantibodies remains to be demonstrated using hinge-specific reagents[48,53,58].

STUDIES WITH DEFINED HAH AUTOANTIBODIES

Precise identification of hinge-region epitopes for HAH and development of highly specific detection

reagents (see specificity section) has led to the identification of HAH autoantibodies as predictors of graft

outcome in kidney transplantation[71,72,73]. Kidney graft recipients with a high pretransplant HAH titer

of the IgG class had a significantly better survival rate than patients with low IgG HAH autoantibodies; a

similar correlation was found in patients with high pretransplant HAH titers of the IgA class. Those

patients with both high IgG and IgA HAH autoantibodies had the best survival rate. The basis of these

correlations is not known, but may be due to an immunosuppressive effect (see above) of the HAH

autoantibodies[71].

A role for HAH autoantibodies (which the authors refer to as antihinge NAbs) in complement

amplification has been elucidated with a combination of in vitro and in vivo studies[13]. Well-

characterized antihinge NAbs were shown to participate in the formation of immune complexes with

antigen-bound F(ab')2 fragments that can stimulate complement amplification via capture of dimeric C3b,

known as a potent C3 convertase precursor (Fig. 2B). The authors suggested that binding of antihinge

NAbs to the F(ab')2 antigen complexes stabilizes the hinge region of the F(ab')2 molecules, and facilitates

binding of dimeric C3b to the known C3b binding site[74] in the CH1 region of the F(ab')2 fragments.

Alternatively, it may be that the antihinge NAbs themselves bind dimeric C3b, leading to active

complement amplification complexes. Excessive complement amplification may trigger the systemic

inflammatory response syndrome (SIRS) following microbial infection. Fumia and colleagues

hypothesize that SIRS may be triggered by F(ab')2 fragments generated from IgG molecules via

proteolytic cleavage, by enzymes released from activated neutrophils or by proteases secreted by

pathogens[13]. The F(ab')2 fragments may then form immune complexes with antigens from the infecting

pathogen and antihinge NAbs that recognize the F(ab')2 hinge region. These complexes can then bind

dimeric C3b, generating complexes that can stimulate complement amplification leading to SIRS.

Page 10: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1162

FIGURE 2. Proposed functional mechanisms for HAH autoantibodies. (A) The Fc region of autoantibodies that

bind to antigen-engaged BCR coligate FcγRIIb, which leads to B-cell suppression (reviewed in Terness et al.[5]).

(B) Antihinge NAbs that bind to immune-complexed F(ab’)2 fragments stiffen the hinge region of the F(ab’)2,

resulting in docking of C3b and complement amplification[13]. (C) HAH autoantibodies bind to F(ab’)2 fragments

on target cells and provide a surrogate Fc domain that can interact with effector immune cells, resulting in target cell

destruction[6].

POSSIBLE ROLE OF HAH AUTOANTIBODIES IN HOST DEFENSE

HAH autoantibodies have been identified in the sera of most normal humans[6,75], raising questions

about their origin and functional significance:

1. How and where are the HAH targets (i.e., proteolyzed antibodies) generated in vivo?

2. What proteases are responsible?

3. What is the normal role(s) of HAH?

4. Are they involved in protection against diseases?

The function(s) of HAH autoantibodies remains to be established. In addition to a possible regulatory

role in immunosuppression (described above), it has been suggested that HAH autoantibodies may play a

role in augmenting host defenses against inflammatory and/or invasive diseases[57]. The targets of HAH

autoantibodies (i.e., fragments containing the hinge region of proteolyzed IgG) have been found in

synovial fluid from rheumatoid arthritis patients[53], breast carcinoma extracts[66], and associated with

bacterial infections such as Streptococcus pyogenes[54] and Pseudomonas aeruginosa[76]. The presence

Page 11: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1163

of proteolyzed IgG associated with both cancerous cells and invasive microorganisms suggests that IgG

proteolysis may be the basis of an immune evasion mechanism. Tumors and many microorganisms

secrete proteases that cleave IgG molecules at defined sites in the hinge region[53,66], potentially

resulting in inactivation or complete removal of the Fc region that normally mediates important host

defense functions, such as ADCC, antibody-dependent cellular phagocytosis (ADCP), and CDC. Support

for this in vivo defense mechanism was provided by in vitro experiments in which proteolysis of purified

IgGs using physiologically relevant proteases targeting the hinge region (e.g., various human matrix

metalloproteinases, GluV8 from Staphylococcus aureus, IdeS from Streptococcus pyogenes) led to

complete loss of ADCC and CDC activities of a monoclonal antibody in vitro with no effect on antigen

binding[66]. The loss of Fc-mediated functions was observed not only when the Fc was completely

removed (both heavy chains cleaved in the hinge region), but even when only a single cleavage event

took place (i.e., only one of the heavy chains was cleaved in the hinge region), and the Fc remained

associated with the antigen binding domains. Importantly, addition of affinity-purified HAH

autoantibodies reactive with F(ab')2 fragments, but not intact IgG, was able to completely restore the lost

ADCC and CDC activity of the proteolytically generated F(ab')2 fragments (Fig. 2C), presumably by

providing a surrogate Fc domain[6]. Thus, a possible in vivo function of HAH autoantibodies is to counter

immune evasion strategies of tumors and invasive microorganisms by restoring key immune system

defense mechanisms.

IMPLICATIONS OF HAH AUTOANTIBODIES FOR USE OF THERAPEUTIC ANTIBODY FRAGMENTS

The prevalence of HAH autoantibodies in the normal human population has the potential to affect the

efficacy of therapeutic monoclonal antibody fragments. The first recombinant monoclonal antibody

product approved in the U.S. was abciximab, a chimeric Fab fragment generated by papain treatment of

IgG. Its molecular target is the GPIIb/IIIa (αIIbβ3) integrin on the surface of human platelets. It was found

that most humans have detectable reactivity to the papain cleavage site in the upper hinge region of

human Fab fragments (i.e., HAH), including abciximab[75]. If pre-existing HAH autoantibodies bound to

abciximab while it was bound to the platelet surface via GPIIb/IIIa during therapy, there was the potential

for platelet clearance mediated by the Fc regions of HAH autoantibodies. In clinical studies, it was found

that although abciximab-coated platelets could bind HAH autoantibodies ex vivo[77], there were few

adverse clinical effects, including acute thrombocytopenia, that could be attributed to HAH

autoantibodies in clinical trials[75]. Interestingly, the pretreatment HAH autoantibody reactivity tended to

increase modestly after abciximab treatment, peaking at the earliest time points tested (1–3 weeks)

compared to typical induced immune responses against the murine variable region of abciximab that

peaked at 4–6 weeks following treatment[75], suggesting that this increased HAH autoantibody reactivity

was unlikely to be a primary induced immune response and was perhaps indicative of an anamnestic

response.

The low rate of acute thrombocytopenia seen in abciximab clinical trials (<2%)[78] may reflect the

relatively low reactivity of HAH autoantibodies targeting the papain cleavage site in the upper hinge

compared to cleavage sites in the lower hinge that generate F(ab')2 fragments[6,57], as discussed

previously. Support for this idea was provided by Yano and coworkers[79], who used a different anti-

GPIIb/IIIa antibody, hC4G1, and its fragments, in a series of studies in cynomolgus monkeys. Following

administration of the pepsin-generated F(ab')2 fragment of hC4G1, severe thrombocytopenia was

observed in five of 18 monkeys. Autoantibodies directed against the hinge region of the F(ab')2 fragment

were detected in most monkeys, and there was an inverse correlation between the level of the antihinge

autoantibodies and platelet counts following F(ab')2 administration, with the five monkeys exhibiting the

highest level of autoantibodies having the most severe platelet depletion. In a separate study,

administration of the Fab fragment of hC4G1 did not lead to thrombocytopenia in monkeys[80].

Page 12: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1164

To avoid the potential for reactivity of future therapeutic antibody fragments with endogenous HAH

autoantibodies, one strategy is to produce recombinant fragments choosing the C-terminal amino acid of

the heavy chain to be a position that is nonreactive with endogenous HAH autoantibodies. Such

nonreactive positions in the hinge were identified using a series of peptide analogs with free C-termini

corresponding to each amino acid position across the IgG1 hinge[6].

CONCLUSIONS

Although the existence of autoantibodies that bind to cryptic epitopes exposed by proteolysis of the IgG

hinge region has been appreciated for some time, the origin and biological relevance of HAH

autoantibodies remains unclear. However, several studies have defined the binding epitopes of HAH

autoantibodies. These specificity studies suggested that there are multiple binding sites in both the upper

and lower hinge region for HAH autoantibodies. Additionally, HAH autoantibodies have been identified

with different isotypes. Taken together, these results suggest that there could be many different antihinge

clones, including NAbs (with germline variable regions), as well as HAH autoantibodies that arose as a

result of immune responses. Several biological mechanisms have been attributed to HAH autoantibodies,

ranging from immunosuppressive effects to augmenting the host immune response. Molecular cloning of

antihinge-specific autoantibodies from B cells should provide a useful tool to define the origin of HAH

autoantibodies by variable region sequence analysis and help to elucidate the biological role of HAH

autoantibodies with defined targets in hinge region.

REFERENCES

1. Baumgarth, N. (2011) The double life of a B-1 cell: self-reactivity selects for protective effector functions. Nat. Rev.

Immunol. 11, 34–46.

2. Lutz, H.U., Binder, C.J., and Kaveri, S. (2009) Naturally occurring auto-antibodies in homeostasis and disease.

Trends Immunol. 30, 43–51.

3. Avrameas, S. (1992) Natural Autoantibodies: Self-Recognition and Physiological Autoimmunity. CRC Press, Boca

Raton, FL.

4. Welschof, M., Terness, P., Kipriyanov, S.M., Stanescu, D., Breitling, F., Dorsam, H., Dubel, S., Little, M., and Opelz,

G. (1997) The antigen-binding domain of a human IgG-anti-F(ab')2 autoantibody. Proc. Natl. Acad. Sci. U. S. A. 94,

1902–1907.

5. Terness, P., Navolan, D., Dufter, C., Welschof, M., and Opelz, G. (2002) Immunosuppressive anti-immunoglobulin

autoantibodies: specificity, gene structure and function in health and disease. Cell. Mol. Biol. (Noisy-le-grand) 48,

271–278.

6. Brezski, R.J., Luongo, J.L., Petrone, D., Ryan, M.H., Zhong, D., Tam, S.H., Schmidt, A.P., Kruszynski, M.,

Whitaker, B.P., Knight, D.M., and Jordan, R.E. (2008) Human anti-IgG1 hinge autoantibodies reconstitute the

effector functions of proteolytically inactivated IgGs. J. Immunol. 181, 3183–3192.

7. Mandy, W.J. (1967) A new serum factor in normal rabbits. 3. Specificity for antigenic determinants uncovered by

papain or pepsin digestion. J. Immunol. 99, 815–824.

8. Mandy, W.J. and Lewis, F.B. (1966) Homoreactant: a new serum factor in "normal" rabbits. Nature 212, 791–792.

9. Terness, P., Kirschfink, M., Navolan, D., Dufter, C., Kohl, I., Opelz, G., and Roelcke, D. (1995) Striking inverse

correlation between IgG anti-F(ab')2 and autoantibody production in patients with cold agglutination. Blood 85, 548–

551.

10. Susal, C., Oberg, H.H., Daniel, V., Dorr, C., Terness, P., Huth-Kuhne, A., Zimmermann, R., and Opelz, G. (1994)

Isotypes and IgG subclasses of anti-Fab antibodies in human immunodeficiency virus-infected hemophilia patients.

Vox Sang. 66, 37–45.

11. Persselin, J.E. and Stevens, R.H. (1985) Anti-Fab antibodies in humans. Predominance of minor immunoglobulin G

subclasses in rheumatoid arthritis. J. Clin. Invest. 76, 723–730.

12. Williams, R.C., Jr., Malone, C.C., Huffman, G.R., Silvestris, F., Croker, B.P., Ayoub, E.M., and Massengill, S.

(1995) Active systemic lupus erythematosus is associated with depletion of the natural generic anti-idiotype (anti-

F(ab')2) system. J. Rheumatol. 22, 1075–1085.

13. Fumia, S., Goede, J.S., Fischler, M., Luginbuhl, A., Frick, S., Fodor, P., and Lutz, H.U. (2008) Human F(ab')(2)-

containing immune complexes together with anti-hinge natural antibodies stimulate complement amplification in vitro

and in vivo. Mol. Immunol. 45, 2951–2961.

Page 13: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1165

14. von Boehmer, H. and Melchers, F. (2010) Checkpoints in lymphocyte development and autoimmune disease. Nat.

Immunol. 11, 14–20.

15. Roy, B., Shukla, S., Lyszkiewicz, M., Krey, M., Viegas, N., Duber, S., and Weiss, S. (2009) Somatic hypermutation

in peritoneal B1b cells. Mol. Immunol. 46, 1613–1619.

16. Sauerborn, M. and Schellekens, H. (2009) B-1 cells and naturally occurring antibodies: influencing the

immunogenicity of recombinant human therapeutic proteins? Curr. Opin. Biotechnol. 20, 715–721.

17. Ehrenstein, M.R. and Notley, C.A. (2010) The importance of natural IgM: scavenger, protector and regulator. Nat.

Rev. Immunol. 10, 778–786.

18. Wardemann, H., Yurasov, S., Schaefer, A., Young, J.W., Meffre, E., and Nussenzweig, M.C. (2003) Predominant

autoantibody production by early human B cell precursors. Science 301, 1374–1377.

19. Nemazee, D.A. and Burki, K. (1989) Clonal deletion of B lymphocytes in a transgenic mouse bearing anti-MHC class

I antibody genes. Nature 337, 562–566.

20. Norvell, A., Mandik, L., and Monroe, J.G. (1995) Engagement of the antigen-receptor on immature murine B

lymphocytes results in death by apoptosis. J. Immunol. 154, 4404–4413.

21. Radic, M.Z., Erikson, J., Litwin, S., and Weigert, M. (1993) B lymphocytes may escape tolerance by revising their

antigen receptors. J. Exp. Med. 177, 1165–1173.

22. Cambier, J.C., Gauld, S.B., Merrell, K.T., and Vilen, B.J. (2007) B-cell anergy: from transgenic models to naturally

occurring anergic B cells? Nat. Rev. Immunol. 7, 633–643.

23. Halverson, R., Torres, R.M., and Pelanda, R. (2004) Receptor editing is the main mechanism of B cell tolerance

toward membrane antigens. Nat. Immunol. 5, 645–650.

24. Meffre, E. and Wardemann, H. (2008) B-cell tolerance checkpoints in health and autoimmunity. Curr. Opin.

Immunol. 20, 632–638.

25. Ahmed, R. and Gray, D. (1996) Immunological memory and protective immunity: understanding their relation.

Science 272, 54–60.

26. McHeyzer-Williams, L.J. and McHeyzer-Williams, M.G. (2005) Antigen-specific memory B cell development. Annu.

Rev. Immunol. 23, 487–513.

27. Rajewsky, K. (1996) Clonal selection and learning in the antibody system. Nature 381, 751–758.

28. Tiller, T., Tsuiji, M., Yurasov, S., Velinzon, K., Nussenzweig, M.C., and Wardemann, H. (2007) Autoreactivity in

human IgG+ memory B cells. Immunity 26, 205–213.

29. Fagarasan, S., Kinoshita, K., Muramatsu, M., Ikuta, K., and Honjo, T. (2001) In situ class switching and

differentiation to IgA-producing cells in the gut lamina propria. Nature 413, 639–643.

30. William, J., Euler, C., Christensen, S., and Shlomchik, M.J. (2002) Evolution of autoantibody responses via somatic

hypermutation outside of germinal centers. Science 297, 2066–2070.

31. Tiller, T., Meffre, E., Yurasov, S., Tsuiji, M., Nussenzweig, M.C., and Wardemann, H. (2008) Efficient generation of

monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J. Immunol.

Methods 329, 112–124.

32. Porter, R.R. (1958) Separation and isolation of fractions of rabbit gamma-globulin containing the antibody and

antigenic combining sites. Nature 182, 670–671.

33. Nisonoff, A., Wissler, F.C., and Lipman, L.N. (1960) Properties of the major component of a peptic digest of rabbit

antibody. Science 132, 1770–1771.

34. Kormeier, L.C., Ing, J.T., and Mandy, W.J. (1968) Specificity of antiglobulin factors in normal human serum reacting

with enzyme digested gamma-G-globulin. J. Immunol. 100, 612–621.

35. Silvestris, F., Williams, R.C., Jr., and Searles, R.P. (1986) Human anti-F(ab')2 antibodies and pepsin agglutinators

react with Fv determinants. Scand. J. Immunol. 23, 499–508.

36. Davey, M.P. and Korngold, L. (1982) Association of anti-F (ab')2 antibodies (pepsin agglutinators) with immune

complexes as determined by enzyme-linked immunosorbent assays. Int. Arch. Allergy Appl. Immunol. 67, 278–283.

37. Osterland, C.K., Harboe, M., and Kunkel, H.G. (1963) Anti-gamma-globulin factors in human sera revealed by

enzymatic splitting of anti-Rh antibodies. Vox Sang. 8, 133–152.

38. Heimer, R., Wolfe, L.D., and Abruzzo, J.L. (1985) The specificity of antibodies to the F(ab')2 fragment of human

IgG. Arthritis Rheum. 28, 562–568.

39. Natvig, J.B. (1970) Human anti-gamma-globulin antibodies specific for gamma G heavy chain subclasses.

Immunology 19, 125–135.

40. Mellbye, O.J. and Natvig, J.B. (1971) Evidence for immune complexes containing antibody to the pepsin site of IgG

in rheumatoid synovial fluids. Clin. Exp. Immunol. 8, 889–899.

41. Waller, M. and Curry, N. (1970) The demonstration of plasmin agglutinators in human sera. Vox Sang. 19, 34–46.

42. Waller, M., Curry, N., and Mallory, J. (1968) Immunochemical and serological studies of enzymatically fractionated

human IgG globulins. I. Hydrolysis with pepsin, papain, ficin and bromelin. Immunochemistry 5, 577–583.

43. Chuba, J.V. (1994) Susceptibility of monoclonal IgG paraproteins to plasmic cleavage using glycerin-stabilized

human plasmin. Biochem. Biophys. Res. Commun. 202, 367–373.

44. Waller, M. and Blaylock, K. (1966) Further studies on the anti-globulin factors in human serum to the pepsin digested

fragment of the Ri anti-Rh antibody. J. Immunol. 97, 438–443.

Page 14: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1166

45. Carrico, R.J., Beagle, R., Usategui-Gomez, M., and Deinhardt, F. (1974) A radioimmunoassay specific for the

antigenic determinants exposed by pepsin digestion of human immunoglobulin G. Immunochemistry 11, 573–579.

46. Ling, N.R. and Drysdale, P. (1981) Antibodies in human sera to F(ab')2 fragments of monoclonal and polyclonal IgG.

Int. Arch. Allergy Appl. Immunol. 66, 459–463.

47. Persselin, J.E. and Stevens, R.H. (1989) Detection and significance of anti-Fab autoantibody. Monogr. Allergy 26,

74–81.

48. Terness, P., Kohl, I., Hubener, G., Battistutta, R., Moroder, L., Welschof, M., Dufter, C., Finger, M., Hain, C., Jung,

M., et al. (1995) The natural human IgG anti-F(ab')2 antibody recognizes a conformational IgG1 hinge epitope. J.

Immunol. 154, 6446–6452.

49. Nasu, H., Chia, D.S., Knutson, D.W., and Barnett, E.V. (1980) Naturally occurring human antibodies to the F(ab')2

portion of IgG. Clin. Exp. Immunol. 42, 378–386.

50. Williams, R.C., Jr., Malone, C.C., Silvestris, F., and Solomon, A. (1995) Molecular localization of human IgG anti-

F(ab')2 reactivity with variable- and constant-region lambda light-chain epitopes. J. Clin. Immunol. 15, 349–362.

51. Diemel, R.V., ter Hart, H.G.J., Derksen, G.J.A., Koenderman, A.H.L., and Aalberse, R.C. (2005) Characterization of

immunoglobulin G fragments in liquid intravenous immunoglobulin products. Transfusion 45, 1601–1609.

52. Gearing, A.J.H., Thorpe, S.J., Miller, K., Mangan, M., Varley, P.G., Dudgeon, T., Ward, G., Turner, C., and Thorpe,

R. (2002) Selective cleavage of human IgG by matrix metalloproteinases, matrilysin and stromelysin. Immunol. Lett.

81, 41–48.

53. Ryan, M.H., Petrone, D., Nemeth, J.F., Barnathan, E., Bjorck, L., and Jordan, R.E. (2008) Proteolysis of purified IgGs

by human and bacterial enzymes in vitro and the detection of specific proteolytic fragments of endogenous IgG in

rheumatoid synovial fluid. Mol. Immunol. 45, 1837–1846.

54. von Pawel-Rammingen, U., Johansson, B.P., and Bjorck, L. (2002) IdeS, a novel streptococcal cysteine proteinase

with unique specificity for immunoglobulin G. EMBO J. 21, 1607–1615.

55. Coussens, L.M., Fingleton, B., and Matrisian, L.M. (2002) Matrix metalloproteinase inhibitors and cancer: trials and

tribulations. Science 295, 2387–2392.

56. Egeblad, M. and Werb, Z. (2002) New functions for the matrix metalloproteinases in cancer progression. Nat. Rev.

Cancer 2, 161–174.

57. Brezski, R.J. and Jordan, R.E. (2010) Cleavage of IgGs by proteases associated with invasive diseases: an evasion

tactic against host immunity? MAbs 2, 212–220.

58. Welschof, M., Reineke, U., Kleist, C., Kipriyanov, S., Little, M., Volkmer-Engert, R., Schneider-Mergener, J., Opelz,

G., and Terness, P. (1999) The antigen binding domain of non-idiotypic human anti-F(ab')2 autoantibodies: study of

their interaction with IgG hinge region epitopes. Hum. Immunol. 60, 282–290.

59. Terness, P., Kohl, I., Hübener, G., Battistutta, R., Moroder, L., Welschof, M., Dufter, C., Finger, M., Hain, C., Jung,

M., and Opelz, G. (1995) The natural human IgG anti-F(ab')2 antibody recognizes a conformational IgG1 hinge

epitope. J. Immunol. 154, 6446–6452.

60. Burton, D.R. (1985) Immunoglobulin G: functional sites. Mol. Immunol. 22, 161–206.

61. Duncan, A.R., Woof, J.M., Partridge, L.J., Burton, D.R., and Winter, G. (1988) Localization of the binding site for the

human high-affinity Fc receptor on IgG. Nature 332, 563–564.

62. Frangione, B. and Milstein, C. (1968) Variations in the S-S bridges of immunoglobins G: interchain disulfide bridges

of gamma G3 myeloma proteins. J. Mol. Biol. 33, 893–906.

63. Schmidt, A., DeRiggi, D., Jordan, R.E., Whitaker, B., Heavner, G.A., and Kruszynski, M. (2009) A synthetic peptide

approach for elucidating the points of natural auto-antibody reactivity to proteolytic fragments of human IgG. Adv.

Exp. Med. Biol. 611, 411–412.

64. Baici, A., Knopfel, M., and Fehr, K. (1982) Cleavage of the four human IgG subclasses with cathepsin G. Scand. J.

Immunol. 16, 487–498.

65. Vincents, B., von Pawel-Rammingen, U., Björck, L., and Abrahamson, M. (2004) Enzymatic characterization of the

streptococcal endopeptidase, IdeS, reveals that it is a cysteine protease with strict specificity for IgG cleavage due to

exosite binding. Biochemistry 43, 15540–15549.

66. Brezski, R.J., Vafa, O., Petrone, D., Tam, S.H., Powers, G., Ryan, M.H., Luongo, J.L., Oberholtzer, A., Knight, D.M.,

and Jordan, R.E. (2009) Tumor-associated and microbial proteases compromise host IgG effector functions by a

single cleavage proximal to the hinge. Proc. Natl. Acad. Sci. U. S. A. 106, 17864–17869.

67. Waller, M., Duma, R.J., Farley, E.D., Jr., and Atkinson, J. (1971) The influence of infection on titres of antiglobulin

antibodies. Clin. Exp. Immunol. 8, 451–459.

68. Susal, C., Daniel, V., Oberg, H.H., Terness, P., Huth-Kuhne, A., Zimmerman, R., and Opelz, G. (1992) Striking

inverse association of IgG-anti-Fab gamma antibodies and CD4 cell counts in patients with acquired

immunodeficiency syndrome (AIDS)/AIDS-related complex. Blood 79, 954–957.

69. Susal, C., Maier, H., Lorenz, K., and Opelz, G. (1994) Association of IgA-anti-Fab autoantibodies with disease stage

in head-and-neck cancer. Int. J. Cancer 57, 47–50.

70. Terness, P., Berteli, A., Susal, C., and Opelz, G. (1992) Regulation of antibody response by an IgG-anti-Ig

autoantibody occurring during alloimmunization. II. Selective inactivation of antigen receptor-occupied B cells.

Transplantation 54, 92–96.

Page 15: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167

1167

71. Susal, C., Kropelin, M., Groth, J., Wiesel, M., May, G., Carl, S., Staehler, G., and Opelz, G. (1996) Protective effect

of autoantibodies against the hinge region of human IgG in kidney graft recipients. Transplantation 62, 1534–1536.

72. Staak, A., Renner, F., Suesal, C., Dietrich, H., Rainer, L., Kamali-Ernst, S., Ernst, W., Padberg, W., Opelz, G., and

Weimer, R. (2006) Immunoglobulin induction therapy in renal transplant recipients: effects on immunoglobulin and

regulatory antibody levels. Transplant. Proc. 38, 3483–3485.

73. Terness, P., Navolan, D., Moroder, L., Siedler, F., Weyher, E., Kohl, I., Dufter, C., Welschof, M., Drugarin, D.,

Schneider, F., and Opelz, G. (1996) A natural IgA-anti-F(ab')2gamma autoantibody occurring in healthy individuals

and kidney graft recipients recognizes an IgG1 hinge region epitope. J. Immunol. 157, 4251–4257.

74. Vidarte, L., Pastor, C., Mas, S., Blazquez, A.B., de los Rios, V., Guerrero, R., and Vivanco, F. (2001) Serine 132 is

the C3 covalent attachment point on the CH1 domain of human IgG1. J. Biol. Chem. 276, 38217–38223.

75. Knight, D.M., Wagner, C., Jordan, R., McAleer, M.F., DeRita, R., Fass, D.N., Coller, B.S., Weisman, H.F., and

Ghrayeb, J. (1995) The immunogenicity of the 7E3 murine monoclonal Fab antibody fragment variable region is

dramatically reduced in humans by substitution of human for murine constant regions. Mol. Immunol. 32, 1271–1281.

76. Fick, R.B., Jr., Baltimore, R.S., Squier, S.U., and Reynolds, H.Y. (1985) IgG proteolytic activity of Pseudomonas

aeruginosa in cystic fibrosis. J. Infect. Dis. 151, 589–598.

77. Curtis, B.R., Swyers, J., Divgi, A., McFarland, J.G., and Aster, R.H. (2002) Thrombocytopenia after second exposure

to abciximab is caused by antibodies that recognize abciximab-coated platelets. Blood 99, 2054–2059.

78. Jordan, R.E., Nakada, M.T., and Weisman, H.F. (1999) Abciximab: the first platelet glycoprotein IIb/IIIa receptor

antagonist. In Biopharmaceuticals, An Industrial Perspective. Walsh, G. and Murphy, B., Eds. Kluwer Academic,

Dordrecht, The Netherlands. pp. 35–71.

79. Yano, S., Kaku, S., Suzuki, K., Terazaki, C., Sakayori, T., Kawasaki, T., Kawamura, K., Sugita, Y., Hoshino, K., and

Masuho, Y. (1995) Natural antibodies against the immunoglobulin F(ab')2 fragment cause elimination of antigens

recognized by the F(ab')2 from the circulation. Eur. J. Immunol. 25, 3128–3133.

80. Kaku, S., Yano, S., Kawasaki, T., Sakai, Y., Suzuki, K., Kawamura, K., Masuho, Y., Satoh, N., Takenaka, T.,

Landolfi, N.F., and Co, M.S. (1996) Comparison of the antiplatelet agent potential of the whole molecule, F(ab)2 and

Fab fragments of humanized anti-GPIIb/IIIa monoclonal antibody in monkeys. Gen. Pharmacol. 27, 435–439.

This article should be cited as follows:

Brezski, R.J., Knight, D.M., and Jordan, R.E. (2011) The origins, specificity, and potential biological relevance of human anti-

IgG hinge autoantibodies. TheScientificWorldJOURNAL 11, 1153–1167. DOI 10.1100/tsw.2011.107.

Page 16: The Origins, Specificity, and Potential Biological Relevance of … · Brezski et al.: Human Anti-IgG Hinge Autoantibodies TheScientificWorldJOURNAL (2011) 11, 1153–1167 1156 regions

Submit your manuscripts athttp://www.hindawi.com

Stem CellsInternational

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

MEDIATORSINFLAMMATION

of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Behavioural Neurology

EndocrinologyInternational Journal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Disease Markers

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

BioMed Research International

OncologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Oxidative Medicine and Cellular Longevity

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

PPAR Research

The Scientific World JournalHindawi Publishing Corporation http://www.hindawi.com Volume 2014

Immunology ResearchHindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Journal of

ObesityJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Computational and Mathematical Methods in Medicine

OphthalmologyJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Diabetes ResearchJournal of

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Research and TreatmentAIDS

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Gastroenterology Research and Practice

Hindawi Publishing Corporationhttp://www.hindawi.com Volume 2014

Parkinson’s Disease

Evidence-Based Complementary and Alternative Medicine

Volume 2014Hindawi Publishing Corporationhttp://www.hindawi.com